Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion

  • Authors:
    • Feng‑Chen Zhu
    • Dian‑Ming Jiang
    • Ming‑Hua Zhang
    • Bo Zhao
    • Chao He
    • Jian Yang
  • View Affiliations

  • Published online on: September 16, 2019     https://doi.org/10.3892/mmr.2019.10687
  • Pages: 4285-4292
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate whether nuclear factor erythroid 2p45‑related factor 2 (Nrf2) overexpression by gene transfer may protect neurons/glial cells and the association between neurons/glial cells and axons in spinal cord injury (SCI). In the present study, Nrf2 recombinant adenovirus (Ad) vectors were constructed. The protein levels of Nrf2 in the nucleus and of the Nrf2‑regulated gene products heme oxygenase‑1 (HO‑1) and NAD (P)H‑quinone oxidoreductase‑1 (NQO1), were detected using western blot analysis in PC12 cells following 48 h of transfection. Furthermore, the expression of Nrf2 was localized using an immunofluorescence experiment, and the expression of Nrf2, HO‑1 and NQO1 were detected using an immunohistochemical experiment in the grey matter of spinal cord in rats. Post‑injury motor behavior was assessed via the Basso, Beattie and Bresnahan (BBB) locomotor scale method. In PC12 cells, subsequent to Ad‑Nrf2 transfection, nuclear Nrf2, HO‑1 and NQO1 levels were significantly increased compared with the control (P<0.01). There was statistically significant changes in the PC12‑Ad‑Nrf2 group [Nrf2 (1.146±0.095), HO‑1 (1.816±0.095) and NQO1 (1.421±0.138)] compared with the PC12‑control group [Nrf2 (0.717±0.055), HO‑1 (1.264±0.081) and NQO1 (0.921±0.088)] and PC12‑Ad‑green fluorescent protein group [Nrf2 (0.714±0.111), HO‑1 (1.238±0.053) and NQO1 (0.987±0.045); P<0.01]. The BBB scores of the rats indicated that they had improved functional recovery following the local injection of Ad‑Nrf2. On the third day following the operation, BBB scores in the adenovirus groups (0.167±0.408) were significantly decreased compared with the SCI group (1±0.894; P<0.05). In the injured section of the spinal cord in the rats, the number of positive cells expressing Nrf2, HO‑1 and NQO1 were raised compared with the control and SCI groups, indicating that the adenovirus vector‑mediated gene transfer of Nrf2 promotes functional recovery following spinal cord contusion in rats.

Introduction

Secondary injury serves a key function in the outcome of patients with spinal cord injury (SCI) (1). It is important to reduce the apoptosis or necrosis of neurons, and maintain the links between neurons/glial cells and axons in SCI (2,3). The mechanism of secondary injury is complex (4). Numerous studies have identified that microvascular perfusion changes, free radical production and lipid peroxidation, necrosis and apoptotic cell death and the dysregulation of ionic homeostasis are able to promote secondary injury following SCI (58). Previous studies have attempted to identify a desirable target which is able to interrupt multi-mechanisms underlying secondary injury.

Nuclear factor erythroid 2p45-related factor 2 (Nrf2) is a member of the Cap ‘n’ Collar basic-leucine-zipper family of transcription factors (9). Under numerous stimuli, Nrf2 translocates from the cytoplasm to the nucleus (10) and sequentially binds to antioxidant response element (ARE) (11). ARE is a promoter element commonly identified in protective genes and its products are involved in reducing oxidative stress, inflammatory damage and reducing the accumulation of toxic metabolites (12). Nrf2 transactivates the expression of a number of cytoprotective enzymes by binding to ARE motifs, including heme oxygenase-1 (HO-1) and NAD(P)H-quinone oxidoreductase-1 (NQO1). These products regulated by the Nrf2 gene, in order to protect the cell from oxidative or xenobiotic damage (1316).

It has been noted that ~7% of the normal number of axons below the injury level are required to mediate meaningful distal neurologic function (17,18). In order to maintain the necessary neurologic functions, 1.4–12% of the total number of axons across the spinal cord injury site are required (1921). Therefore, even small increases in neuroprotection may affect functionally relevant neurologic recovery and thus is important for SCI patients (22). However, neurons have low antioxidant abilities and are highly sensitive to oxidative stress, therefore, increased levels of reactive oxygen species (ROS) easily induces neuron damage (23,24). The Nrf2-ARE pathway in central nervous system (CNS) injury serves a protective function (25). It has been proven that Nrf2 serves pivotal functions in the cell, which may defend against the oxidative stress of traumatic brain injury (TBI) in rats or mice, and decrease the severity of neurological deficit. On the contrary, Nrf2 knockout increased the severity of TBI, even with the use of an Nrf2 inducer (26,27). Furthermore, the disruption of Nrf2 may upregulate the activity of nuclear factor-κβ and proinflammatory cytokines following TBI or SCI in mice (28,29). The aim of the present study was to investigate whether Nrf2 gene transfer overexpression can protect neurons/glial cells, and the association between neurons/glial cells and axons during SCI.

Materials and methods

Experimental overview

Gene transfer has been widely used for experimental research (30). However, to the best of our knowledge, Nrf2 gene transfer to TBI and SCI have not been reported. In the present study, Nrf2 recombinant adenovirus vectors were constructed that were then transfected into PC12 cells and locally injected into SCI in rats. The protein levels of Nrf2 in the nucleus and the Nrf2-regulated gene expression of HO-1 and NQO1 were detected using western blot analysis in PC12 cells following 48 h of transfection. Furthermore, the expression of Nrf2 was localized by using immunofluorescence and the expression of Nrf2, HO-1 and NQO1 were detected using immunohistochemistry in the grey matter of the spinal cord in rats. Post-injury motor behavior was assessed via the Basso, Beattie and Bresnahan (BBB) locomotor scale method.

Cell line

PC12 cell line (a neuron model) was provided by Department of Neurology of the First Affiliated Hospital of Chongqing Medical University (Chongqing, China) and cultured in RPMI 1640 medium (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) with 10% calf serum (HyClone; GE Healthcare Life Sciences), 5% horse serum (HyClone; GE Healthcare Life Sciences), 100 U/ml penicillin and 100 µg/ml streptomycin. Cells were cultured in a humidified atmosphere incubator at 5% CO2 and 37°C, and the medium was changed every other day.

Constructing recombinant adenoviral vectors

The adenovirus shuttle plasmids pAV-MCMV-green fluorescent protein (GFP)-Nrf2 and pAV-MCMV-GFP were purchased from Microbix Biosystems (Mississauga, ON, Canada). Recombinant adenoviral vectors were generated by using the Admax Cre-lox system (Microbix Biosystems, Inc.). The adenovirus was propagated in 293 cells (Health Science Research Resources Bank Osaka, Japan). and purified by CsCl2 density gradient centrifugation (40,000 × g, 2 h, 4°C). Virus titers were determined using plaque assays. For PC12 cell transfection, 2 ml PC12 cells (1×105/ml) supernatant was incubated at 37°C in 6-well plates overnight. Then they were infected for 20 min at 37°C with viral supernatant containing vectors at a multiplicity of infection of 100 in the presence of 8 µg/ml polybrene. PC12 cells were divided into three groups: PC12-Control (no virus infection), PC12-Ad-Nrf2 group (Ad-Nrf2 infection) and PC12-Ad-GFP group (Ad-GFP infection).

Animal preparation

A total of 100 adult (5 weeks) male and female Sprague-Dawley rats of Specific-pathogen free (SPF) (180–250 g; Chongqing Medical University, Chongqing, China) were housed under a room temperature and humidity (24°C and 50%) on a 12 h light-dark cycle with ad libitum access to food and water. All experimental procedures were performed in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals (NIH publication no. 80-23, revised 1996), and the number of animals used and their suffering were minimized. Ethical approval was provided by the Yongchuan Hospital of Chongqing Medical University (Chongqing, China). Using a random number table, rats were divided into: i) a Sham-operated group (n=25), ii) a SCI group (n=25), iii) an Ad-Nrf2 group (n=25), and iv) an Ad-GFP group (n=25). On the 1st, 3rd, 7th, 14th and 28th day subsequent to surgery, 5 surviving rats were selected from each group for further experimental study.

The extradural compression of the modified Allen's method was used to produce the SCI animal model (31). Briefly; rats were anesthetized intraperitoneally with chloral hydrate (300 mg/kg) and underwent a laminectomy to expose the dorsal portion of spinal cord from T8 to T10 levels. Moderate or severe contusion injury was performed with a weight-drop device by dropping a 10 g rod (3 mm in diameter, 5 cm in height with injury pulse 10×5 gcf). Within 30 min following the injury, 1 µl adenoviral vector, diluted to 5×1010 pfu/ml prior to use, was injected into the 2 mm spinal cord stump from the wound site vertically into each stump at a depth of 0.8 mm using a Hamilton micro-injector. The injection rate was slow in order to minimize damage to the spinal cord. Each animal was injected with a total of 5×108 pfu viruses. A constant body temperature was maintained with an overhead heating lamp during the experiment. In the Sham-operated group, the rats underwent the same laminectomy procedure but no trauma was produced. The GFP expression of PC12 cells were detected at 48 h after virus transfection with fluorescence microscopy.

Western blot analysis

The proteins were extracted using a commercial kit according to the manufacturer's protocol (NBP2-37853, Novus Biologicals), and the protein expression of Nrf2, HO-1 and NQO1 in a neuron model (PC12 cells) were analyzed using western blot analysis. Briefly, 50 µg of protein extracts determined by Pierce BCA Protein Assay Kit (23227, Thermo Fisher Scientific, Inc.) were separated on 15% SDS-PAGE and then were electrotransferred to a polyvinylidene difluoride filter (PVDF, EMD Millipore, Billerica, MA, USA). The wet electroblotting (using Mini Trans-Blot Module, 1703935, Bio-Rad Laboratories, Inc.) was performed at constant voltage (20 V) for 1 h at 4°C with CAPS based transfer buffer (10 mM CAPS, pH 11, 10% methanol) Then the membranes were blocked with 5% nonfat milk for 1 h at room temperature and incubated with primary antibodies anti-Nrf2 (ab89443, 1:500; Abcam), anti-HO-1 (CL5275, 1:500; Abcam), anti-NQO1 antibodies (ab28947, 1:500; Abcam) and anti β-actin (mAbcam 8226, 1:2,000; Abcam) overnight at 4°C, followed by anti-goat immunoglobulin G horseradish peroxidase-conjugated secondary antibodies (sc-2354, 1:5,000; Santa Cruz Biotechnology, Inc.) for 2 h at room temperature. Following rinsing with a buffer, the protein bands were visualized using an enhanced chemiluminescence kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Film signals were digitally quantified by internal control β-actin using Quantity 4.6.2 software (Bio-Rad Laboratories, Inc.).

BBB locomotor scale

The BBB locomotor scale method was used to assess the hind limb functional improvement of treated animals with spinal cord contusion (32). At 1, 3, 7, 14 and 28 days subsequent to surgery, behavioral analysis was performed based on movement of the hind limb, weight support, forelimb-hind limb coordination and trunk stability.

GFP detection

All animals were perfused transcardially with 4% paraformaldehyde. The cords, ~2 cm in length with the contused site at the center of the sample, were segmented. The parasagittal ice-frozen spinal cord sections (~500 µm away from the sagittal plane, with a thickness of 10 µm) were prepared to detect GFP expression with fluorescent microscopy.

Immunohistochemistry

Five samples from each group on the 3rd postoperative day were fixed with 4% paraformaldehyde for 48 h at room temperature, embedded in paraffin and cross-sections (thickness, 10 µm) were prepared. After deparaffinization and dehydration, sections were boiled in Tris-EDTA buffer (10 mM Tris Base, 1 mM EDTA Solution, 0.05% Tween-20, pH 9.0) for antigen unmasking, followed by extensive washing with PBS. Sections were subsequently incubated with 3% H2O2 for 10 min, and then rinsed with PBS for three times. Sections were blocked with 2% goat serum albumin in PBS for 20 min and incubated with mouse anti-Nrf2, anti-HO-1 and anti-NQO1 antibodies (all at a dilution of 1:100) overnight at 4°C. Sections incubated without a primary antibody were used as negative controls. After washing 3 times with PBS, sections were incubated with Goat anti-Mouse IgG Secondary Antibody [HRP (Horseradish Peroxidase), HAF007, 1;1,000, Novus Biologicals] for 10 min at 37°C. After washing with PBS, peroxidase was stained with Mouse specific HRP/DAB (ABC) Detection IHC kit (ab64259; Abcam) and viewed under a light microscope (magnification, ×400,; Olympus BX50; Olympus Corporation). The positive cells were counted in five different fields of view in the gray matter in five random sections of each rat. All images were captured using Sim PCI 6.0 (Compix Media, Inc.).

Statistical analysis

Data were analyzed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA) and were presented as the mean ± standard error of the mean. Statistical differences were measured using a one-way analysis of variance and Bonferroni's test to compare the differences between groups. P<0.05 was considered to indicate a statistically significant difference.

Results

PC12 cell data

GFP expression in PC12 cells was observed 48 h subsequent to adenovirus transfection. It is likely that Nrf2 is able to affect PC12 proliferation and axonal growth, so the morphology is different between Fig. 1A-C and between Fig. 1D-F (33). The expression of GFP following Ad-Nrf2 transfection was mainly located in the nucleus, and its expression in the cytoplasm was observed following Ad-GFP transfection (Fig. 1A-F).

Western blot analysis was performed to evaluate the protein levels of nuclear Nrf2 (57 kDa), HO-1 (32 kDa) and NQO1 (30 kDa) in whole cells. Subsequent to Ad-Nrf2 transfection, nuclear Nrf2, HO-1 and NQO1 were significantly increased compared with the control (P<0.01; Fig. 1G-I). There was statistically significant changes in the PC12-Ad-Nrf2 group [Nrf2 (1.146±0.095), HO-1 (1.816±0.095) and NQO1 (1.421±0.138)] compared with the PC12-Control [Nrf2 (0.717±0.055), HO-1 (1.264±0.081) and NQO1 (0.921±0.088)] and PC12-Ad-GFP group [Nrf2 (0.714±0.111), HO-1 (1.238±0.053) and NQO1 (0.987±0.045); (P<0.01)].

BBB Locomotor scale results

The animal behavioral analysis was performed at 1, 3, 7, 14 and 28 days following the operation. In the Sham-operated group, BBB scores between day 1 and day 28 did not decrease significantly. On the 3rd day following the operation, BBB scores in the Ad-Nrf2 group (0.167±0.408) exhibited a significantly decreased comparing with the SCI group (1±0.894; P<0.05). On the 7th day following the operation, the BBB scores in the Ad-Nrf2 group (2.333±0.516) were significantly increased compared with the Ad-GFP group (0.9±0.21; P<0.05). On the 14th and 28th day, there was a statistically significant increase in the Ad-Nrf2 group (day 14, 9.833±1.17; day 28, 14±2.608) compared with the SCI group (day 14, 8±1.265; day 28, 11.167±1.901) and the Ad-GFP group (day 14, 7.167±1.17; day 28, 10.333±1.633; P<0.05; Fig. 2).

Gene transfer efficacy

To identify the efficacy of the gene transfer of Ad-Nrf2 and Ad-GFP, the ice-frozen parasagittal sections of the spinal cords, ~2 cm in length with the contused site at the center of the sample, were detected using a fluorescence microscope. Green fluorescence was detected in the spinal cords, primarily near the injected sections, on the 1st day following SCI. The prevalence of the fluorescence expression was observed on the 3rd day following SCI and the contused parts also exhibited GFP expression. From that point onwards, the fluorescence expression decreased gradually (Fig. 3A-E).

Immunohistochemical study

The expression levels of Nrf2, HO-1 and NQO1 in grey matter were localized and analyzed using an immunohistochemical experiment on the 3rd day following the operation. Very few cells were positive for Nrf2, HO-1 and NQO1 in the Sham-operated group. Nrf2, HO-1 and NQO1 immunoreactivity was present in neurons and glial cells following SCI. In the Ad-Nrf2 group, the positive neurons and glial cells for Nrf2, HO-1 and NQO1 were significantly increased compared with the control and SCI groups (P<0.01; Fig. 4). It should be noted that due to the poor homogeneity of spinal cord tissue, the tissue specimens from different rats ought to be treated carefully at the time of fixing and the section orientated to reduce the morphological effects.

Discussion

The leucine zipper transcription factor Nrf2 is a major component of ARE-driven gene expression (33). Oligonucleotide microarray analysis has indicated that Nrf2 is necessary in combating electrophiles and ROS (23,3437). It serves a key function in protecting cells from oxidative stress. Nrf2 may protect the liver from acetaminophen-induced injury (38) and the lung from butylated hydroxytoluene-induced toxicity (39). Cho et al (40) demonstrated that disruption of Nrf2 significantly enhanced pulmonary sensitivity and responsivity to hyperoxic challenge. Compared with Nrf2+/+ astrocytes, one previous study confirmed that Nrf2−/− primary astrocytes are more susceptible to oxidative stress and inflammation (37). Nrf2+/+ astrocytes pretreated with t-butylhydroquinone induce Nrf2 nuclear translocation, resulting in the coordinated upregulation of ARE-driven genes and attenuation of H2O2 and platelet-activating factor-induced cell death (37).

PC12 cells (adrenal pheochromocytoma) was originally isolated from tumors in the rat adrenal medulla in 1976 (41). They resemble the phenotype of sympathetic ganglion neurons upon differentiation with nerve growth factor (NGF) and may be subcultured indefinitely. The PC12 cell line is traceable to a pheochromocytoma from the rat adrenal medulla (4245). It has been used as the classical neuronal cell model due to its ability to acquire the features of sympathetic neurons (46,47). PC12 cells have been used to investigate the cellular mechanisms by which prion protein fragments cause neuronal dysfunction (48), the nerve injury-induced neuropathic pain model (49), the nitric oxide-induced neurotoxicity model (50) and NGF inducing the differentiation of PC12 cells by functioning through the tropomyosin receptor kinase A receptor (51).

In the present study, the Ad-Nrf2 gene was successfully transferred into PC12 cells and the spinal cord (by local injection). Furthermore, the protein levels of nuclear Nrf2 and its regulated gene products, HO-1 and NQO1, were significantly increased in PC12 cells compared with the control, P<0.01), and the number of positive cells for Nrf2, HO-1 and NQO1 were promoted in the neurons/glial cells of the spinal cord grey matter. The function of the hind limb in SCI rats was significantly improved following Ad-Nrf2 gene transfer compared with the SCI group (P<0.05). According to these results, it was postulated that the gene transfer of Nrf2 was able to alleviate SCI and promote the functional recovery of the injured spinal cord. In view of the fact that neurons are more susceptible than glial cells to oxidative stress in Ad-Nrf2 group (23,52), it may be considered that the Nrf2-ARE pathway may serve a protective function in the pathological process of SCI.

Nrf2 mediates a group of cytoprotective enzymes. It is believed to be the key regulator in CNS diseases by inducing the expression of a group of antioxidant and detoxification enzymes (26,53). In the present study, it was revealed that SCI-induction significantly increased the number of positive cells for Nrf2, HO-1 and NQO1 in grey matter neurons compared with the control (P<0.01), suggesting that the Nrf2-ARE pathway was activated. Such phenomena have additionally been observed in TBI (54).

Increasingly, evidence suggests that a group of cytoprotective enzymes mediated by the Nrf2-ARE pathway serve a pivotal role in antioxidant, anti-inflammation and detoxification functions, including HO-1 and NQO1 (55,56). HO-1 produces biliverdin and reduces bilirubin to reduce ROS production. The expression of ferritin (HO-1 dependent) may prevent the conversion of H2O2 to hydroxyl radicals by the Fenton reaction (57). NQO1 catalyzes the double-electron reduction and detoxification of quinones and their derivatives, therefore protecting cells from the harmful effects of quinones and their associated compounds (58). In SCI, it is considered important to reduce the neurons apoptosis or necrosis and maintain the associations between neurons and axons. However, neurons have a low antioxidant capacity and are highly sensitive to oxidative stress (59). It is reported that Nrf2-mediated neuroprotection is conferred primarily by glia (23). SCI induces an increase in oxidative stress and simultaneously causes glial dysfunction (60). It is therefore important to activate the remaining functional glia and neuronal self-protection. The present study hypothesized that the gene-transfer of Nrf2 may promote the Nrf2-ARE pathway to reduce neuron necrosis or apoptosis, particularly by self-protection. The results revealed that Ad-Nrf2 increased nuclear Nrf2, HO-1 and NQO1 expression in PC12 cells and neurons in the contusion site of SCI, and hind limb functional recovery was also observed. Certain phase II enzyme inducers (activating Nrf2), even fibroblast growth factor-1, have demonstrated neuroprotective effects on motor neuron survival in traumatic SCI (6163). Genetic ablation of the transcription repressor Bach1, a transcriptional repressor of the HO-1 gene, may substantially increase HO-1 expression and cytoprotection against SCI (64). The present study did not successfully isolate the spinal cord neurons of adult rats. PC12 is a tumor cell, which is still different from the neurons themselves. This is the main limitation of the present experiment.

Therefore in conclusion, Nrf2 gene transfer may be a direct method of protecting neurons/glial cells in SCI. Although local injection may injure nerve cells, this method works and will still benefit clinical treatment. Nrf2-adenovirus-mediated in vivo gene transfer may promote functional recovery following spinal cord contusion.

Acknowledgments

Not applicable.

Funding

The present study was supported by the Scientific and Technological Research Program of Chongqing Municipal Education Commission (grant no. KJ130315) and the Recruited Talent Supporting Program of Yongchuan Hospital Affiliated Chongqing Medical University (grant no. YJYJ20120004).

Availability of data and materials

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

Authors' contributions

FCZ was responsible for the study design, literature research, experiments, manuscript preparation, editing and review. DMJ was responsible for the conception of the study and the guarantor of integrity of the entire study. MHZ was responsible for the definition of intellectual content and acquisition of data. BZ was responsible for data acquisition. CH was responsible for data analysis. JY was responsible for statistical analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experimental procedures were performed in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals (NIH publication no. 80-23, revised 1996), and the number of animals used and their suffering were minimized. Ethical approval was provided by the Yongchuan Hospital of Chongqing Medical University (Chongqing, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Oyinbo CA: Secondary injury mechanisms in traumatic spinal cord injury: A nugget of this multiply cascade. Acta Neurobiol Exp (Wars). 71:281–299. 2011.PubMed/NCBI

2 

Gwak YS, Hulsebosch CE and Leem JW: Neuronal-glial interactions maintain chronic neuropathic pain after spinal cord injury. Neural Plast. 2017:24806892017. View Article : Google Scholar : PubMed/NCBI

3 

Alizadeh A, Dyck SM and Karimi-Abdolrezaee S: Traumatic spinal cord injury: An overview of pathophysiology, models and acute injury mechanisms. Front Neurol. 10:2822019. View Article : Google Scholar : PubMed/NCBI

4 

Bylicky MA, Mueller GP and Day RM: Mechanisms of endogenous neuroprotective effects of astrocytes in brain injury. Oxid Med Cell Longev. 2018:65010312018. View Article : Google Scholar : PubMed/NCBI

5 

Kwon BK, Tetzlaff W, Grauer JN, Beiner J and Vaccaro AR: Pathophysiology and pharmacologic treatment of acute spinal cord injury. Spine J. 4:451–464. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Kwon BK, Oxland TR and Tetzlaff W: Animal models used in spinal cord regeneration research. Spine (Phila Pa 1976). 27:1504–1510. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Lukácová N, Halát G, Chavko M and Marsala J: Ischemia-reperfusion injury in the spinal cord of rabbits strongly enhances lipid peroxidation and modifies phospholipid profiles. Neurochem Res. 21:869–873. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Emery E, Aldana P, Bunge MB, Puckett W, Srinivasan A, Keane RW, Bethea J and Levi AD: Apoptosis after traumatic human spinal cord injury. J Neurosurg. 89:911–920. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Andrews NC, Erdjument-Bromage H, Davidson MB, Tempst P and Orkin SH: Erythroid transcription factor NF-E2 is a haematopoietic-specific basic-leucine zipper protein. Nature. 362:722–728. 1993. View Article : Google Scholar : PubMed/NCBI

10 

Motohashi H, Katsuoka F, Engel JD and Yamamoto M: Small Maf proteins serve as transcriptional cofactors for keratinocyte differentiation in the Keap1-Nrf2 regulatory pathway. Proc Natl Acad Sci USA. 101:6379–6384. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Jain AK, Bloom DA and Jaiswal AK: Nuclear import and export signals in control of Nrf2. J Biol Chem. 280:29158–29168. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al: An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 236:313–322. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Jazwa A and Cuadrado A: Targeting heme oxygenase-1 for neuroprotection and neuroinflammation in neurodegenerative diseases. Curr Drug Targets. 11:1517–1531. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Niture SK, Kaspar JW, Shen J and Jaiswal AK: Nrf2 signaling and cell survival. Toxicol Appl Pharmacol. 244:37–42. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Tanito M, Agbaga MP and Anderson RE: Upregulation of thioredoxin system via Nrf2-antioxidant responsive element pathway in adaptive-retinal neuroprotection in vivo and in vitro. Free Radic Biol Med. 42:1838–1850. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Du H, Ma L, Chen G and Li S: The effects of oxyresveratrol abrogates inflammation and oxidative stress in rat model of spinal cord injury. Mol Med Rep. 17:4067–4073. 2018.PubMed/NCBI

17 

Kaelan C, Jacobsen P, Morling P and Kakulas BA: A quantitative study of motoneurons and cortico-spinal fibers related to function in human spinal cord injury (SCI). Paraplegia. 27:1531989.

18 

Kakulas BA: The applied neuropathology of human spinal cord injury. Spinal Cord. 37:79–88. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Blight AR: Cellular morphology of chronic spinal cord injury in the cat: Analysis of myelinated axons by line-sampling. Neuroscience. 10:521–543. 1983. View Article : Google Scholar : PubMed/NCBI

20 

Eidelberg E, Straehley D, Erspamer R and Watkins CJ: Relationship between residual hindlimb-assisted locomotion and surviving axons after incomplete spinal cord injuries. Exp Neurol. 56:312–322. 1977. View Article : Google Scholar : PubMed/NCBI

21 

Fehlings MG and Tator CH: The relationships among the severity of spinal cord injury, residual neurological function, axon counts, and counts of retrogradely labeled neurons after experimental spinal cord injury. Exp Neurol. 132:220–228. 1995. View Article : Google Scholar : PubMed/NCBI

22 

Delamarter RB and Coyle J: Acute management of spinal cord injury. J Am Acad Orthop Surg. 7:166–175. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Shih AY, Johnson DA, Wong G, Kraft AD, Jiang L, Erb H, Johnson JA and Murphy TH: Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress. J Neurosci. 23:3394–3406. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Xu W, Chi L, Xu R, Ke Y, Luo C, Cai J, Qiu M, Gozal D and Liu R: Increased production of reactive oxygen species contributes to motor neuron death in a compression mouse model of spinal cord injury. Spinal Cord. 43:204–213. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Adibhatla RM and Hatcher JF: Lipid oxidation and peroxidation in CNS health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 12:125–169. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Hong Y, Yan W, Chen S, Sun CR and Zhang JM: The role of Nrf2 signaling in the regulation of antioxidants and detoxifying enzymes after traumatic brain injury in rats and mice. Acta Pharmacol Sin. 31:1421–1430. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Jin W, Wang H, Yan W, Zhu L, Hu Z, Ding Y and Tang K: Role of Nrf2 in protection against traumatic brain injury in mice. J Neurotrauma. 26:131–139. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Jin W, Wang H, Yan W, Xu L, Wang X, Zhao X, Yang X, Chen G and Ji Y: Disruption of Nrf2 enhances upregulation of nuclear factor-kappaB activity, proinflammatory cytokines, and intercellular adhesion molecule-1 in the brain after traumatic brain injury. Mediators Inflamm. 2008:7251742008. View Article : Google Scholar : PubMed/NCBI

29 

Mao L, Wang H, Qiao L and Wang X: Disruption of Nrf2 enhances the upregulation of nuclear factor-kappaB activity, tumor necrosis factor-α, and matrix metalloproteinase-9 after spinal cord injury in mice. Mediators Inflamm. 2010:2383212010. View Article : Google Scholar : PubMed/NCBI

30 

Kay MA: State-of-the-art gene-based therapies: The road ahead. Nat Rev Genet. 12:316–328. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Tasdemiroglu E and Tibbs PA: Long-term follow-up results of thoracolumbar fractures after posterior instrumentation. Spine (Phila Pa 1976). 20:1704–1708. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Basso DM, Beattie MS and Bresnahan JC: A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma. 12:1–21. 1995. View Article : Google Scholar : PubMed/NCBI

33 

Tonelli C, Chio IIC and Tuveson DA: Transcriptional regulation by Nrf2. Antioxid Redox Signal. 29:1727–1745. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Moi P, Chan K, Asunis I, Cao A and Kan YW: Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proc Natl Acad Sci USA. 91:9926–9930. 1994. View Article : Google Scholar : PubMed/NCBI

35 

Fan Z, Wirth AK, Chen D, Wruck CJ, Rauh M, Buchfelder M and Savaskan N: Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 6:e3712017. View Article : Google Scholar : PubMed/NCBI

36 

Lee JM, Calkins MJ, Chan K, Kan YW and Johnson JA: Identification of the NF-E2-related factor-2-dependent genes conferring protection against oxidative stress in primary cortical astrocytes using oligonucleotide microarray analysis. J Biol Chem. 278:12029–12038. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Thimmulappa RK, Mai KH, Srisuma S, Kensler TW, Yamamoto M and Biswal S: Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray. Cancer Res. 62:5196–5203. 2002.PubMed/NCBI

38 

Chan K, Han XD and Kan YW: An important function of Nrf2 in combating oxidative stress: Detoxification of acetaminophen. Proc Natl Acad Sci USA. 98:4611–4616. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Chan K and Kan YW: Nrf2 is essential for protection against acute pulmonary injury in mice. Proc Natl Acad Sci USA. 96:12731–12736. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Cho HY, Jedlicka AE, Reddy SP, Kensler TW, Yamamoto M, Zhang LY and Kleeberger SR: Role of NRF2 in protection against hyperoxic lung injury in mice. Am J Respir Cell Mol Biol. 26:175–182. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Greene LA and Tischler AS: Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA. 73:2424–2428. 1976. View Article : Google Scholar : PubMed/NCBI

42 

Uceda G, Artalejo AR, López MG, Abad F, Neher E and García AG: Ca(2+)-activated K+ channels modulate muscarinic secretion in cat chromaffin cells. J Physiol. 454:213–230. 1992. View Article : Google Scholar : PubMed/NCBI

43 

Zhou Z and Neher E: Calcium permeability of nicotinic acetylcholine receptor channels in bovine adrenal chromaffin cells. Pflugers Arch. 425:511–517. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Nooney JM, Peters JA and Lambert JJ: A patch clamp study of the nicotinic acetylcholine receptor of bovine adrenomedullary chromaffin cells in culture. J Physiol. 455:503–527. 1992. View Article : Google Scholar : PubMed/NCBI

45 

Horrigan FT and Bookman RJ: Releasable pools and the kinetics of exocytosis in adrenal chromaffin cells. Neuron. 13:1119–1129. 1994. View Article : Google Scholar : PubMed/NCBI

46 

Westerink RH and Ewing AG: The PC12 cell as model for neurosecretion. Acta Physiol (Oxf). 192:273–285. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Hu R, Cao Q, Sun Z, Chen J, Zheng Q and Xiao F: A novel method of neural differentiation of PC12 cells by using Opti-MEM as a basic induction medium. Int J Mol Med. 41:195–201. 2018.PubMed/NCBI

48 

Taylor SC, Green KN, Smith IF and Peers C: Prion protein fragment 106–126 potentiates catecholamine secretion from PC-12 cells. Am J Physiol Cell Physiol. 281:C1850–C1857. 2001. View Article : Google Scholar : PubMed/NCBI

49 

Shao J, Cao J, Wang J, Ren X, Su S, Li M, Li Z, Zhao Q and Zang W: MicroRNA-30b regulates expression of the sodium channel Nav1.7 in nerve injury-induced neuropathic pain in the rat. Mol Pain. 12:2016. View Article : Google Scholar : PubMed/NCBI

50 

Zheng W, Chong CM, Wang H, Zhou X, Zhang L, Wang R, Meng Q, Lazarovici P and Fang J: Artemisinin conferred ERK mediated neuroprotection to PC12 cells and cortical neurons exposed to sodium nitroprusside-induced oxidative insult. Free Radic Biol Med. 97:158–167. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Liu L, Sun T, Xin F, Cui W, Guo J and Hu J: Nerve growth factor protects against alcohol-induced neurotoxicity in PC12 cells via I3K/Akt/mTOR pathway. Alcohol Alcohol. 52:12–18. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Gilgun-Sherki Y, Melamed E and Offen D: Oxidative stress induced-neurodegenerative diseases: The need for antioxidants that penetrate the blood brain barrier. Neuropharmacology. 40:959–975. 2001. View Article : Google Scholar : PubMed/NCBI

53 

van Muiswinkel FL and Kuiperij HB: The Nrf2-ARE signalling pathway: Promising drug target to combat oxidative stress in neurodegenerative disorders. Curr Drug Targets CNS Neurol Disord. 4:267–281. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Yan W, Wang HD, Hu ZG, Wang QF and Yin HX: Activation of Nrf2-ARE pathway in brain after traumatic brain injury. Neurosci Lett. 431:150–154. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Reisman SA, Buckley DB, Tanaka Y and Klaassen CD: CDDO-Im protects from acetaminophen hepatotoxicity through induction of Nrf2-dependent genes. Toxicol Appl Pharmacol. 236:109–114. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Zhang Y, Guan L, Wang X, Wen T, Xing J and Zhao J: Protection of chlorophyllin against oxidative damage by inducing HO-1 and NQO1 expression mediated by PI3K/Akt and Nrf2. Free Radic Res. 42:362–371. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Jansen T and Daiber A: Direct antioxidant properties of bilirubin and biliverdin. Is there a role for biliverdin reductase? Front Pharmacol. 3:302012.

58 

Gaikwad A, Long DJ II, Stringer JL and Jaiswal AK: In vivo role of NAD(P)H: Quinone oxidoreductase 1 (NQO1) in the regulation of intracellular redox state and accumulation of abdominal adipose tissue. J Biol Chem. 276:22559–22564. 2001. View Article : Google Scholar : PubMed/NCBI

59 

Chen X, Guo C and Kong J: Oxidative stress in neurodegenerative diseases. Neural Regen Res. 7:376–385. 2012.PubMed/NCBI

60 

Santos-Nogueira E, López-Serrano C, Hernández J, Lago N, Astudillo AM, Balsinde J, Estivill-Torrús G, de Fonseca FR, Chun J and López-Vales R: Activation of lysophosphatidic acid receptor type 1 contributes to pathophysiology of spinal cord injury. J Neurosci. 35:10224–10235. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Liu XY, Li CY, Bu H, Li Z, Li B, Sun MM, Guo YS, Zhang L, Ren WB, Fan ZL, et al: The neuroprotective potential of phase II enzyme inducer on motor neuron survival in traumatic spinal cord injury in vitro. Cell Mol Neurobiol. 28:769–779. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Sun MM, Bu H, Li B, Yu JX, Guo YS and Li CY: Neuroprotective potential of phase II enzyme inducer diallyl trisulfide. Neurol Res. 31:23–27. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Vargas MR, Pehar M, Cassina P, Martínez-Palma L, Thompson JA, Beckman JS and Barbeito L: Fibroblast growth factor-1 induces heme oxygenase-1 via nuclear factor erythroid 2-related factor 2 (Nrf2) in spinal cord astrocytes: Consequences for motor neuron survival. J Biol Chem. 280:25571–25579. 2005. View Article : Google Scholar : PubMed/NCBI

64 

Kanno H, Ozawa H, Dohi Y, Sekiguchi A, Igarashi K and Itoi E: Genetic ablation of transcription repressor Bach1 reduces neural tissue damage and improves locomotor function after spinal cord injury in mice. J Neurotrauma. 26:31–39. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 20 Issue 5

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
Zhu FC, Jiang DM, Zhang MH, Zhao B, He C and Yang J: Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion. Mol Med Rep 20: 4285-4292, 2019
APA
Zhu, F., Jiang, D., Zhang, M., Zhao, B., He, C., & Yang, J. (2019). Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion. Molecular Medicine Reports, 20, 4285-4292. https://doi.org/10.3892/mmr.2019.10687
MLA
Zhu, F., Jiang, D., Zhang, M., Zhao, B., He, C., Yang, J."Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion". Molecular Medicine Reports 20.5 (2019): 4285-4292.
Chicago
Zhu, F., Jiang, D., Zhang, M., Zhao, B., He, C., Yang, J."Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion". Molecular Medicine Reports 20, no. 5 (2019): 4285-4292. https://doi.org/10.3892/mmr.2019.10687