Open Access

Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis

  • Authors:
    • Lie Xiong
    • Jingruo Zhang
    • Hanqiang Shi
    • Gaofeng Zhu
    • Xiaoyan Ji
    • Mengjiao Li
    • Ping Zhu
    • Kaitao Luo
  • View Affiliations

  • Published online on: November 23, 2022     https://doi.org/10.3892/etm.2022.11724
  • Article Number: 25
  • Copyright: © Xiong 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

TNFα‑induced protein 1 (TNFAIP1) serve a role in neurovascular disease. However, the potential role and molecular mechanism of TNFAIP1 in cerebral ischemia‑reperfusion (I/R) remains elusive. In the present study, reverse transcription‑quantitative PCR and western blotting were used to assess TNFAIP1 mRNA and protein expression levels in PC12 cells. Furthermore, using Cell Counting Kit‑8, flow cytometry and western blotting, cell viability and apoptosis were evaluated. Oxidative stress was evaluated using DCFH‑DA staining and ELISA was used for assessment of inflammatory factors. Expression of components in the nuclear factor erythroid 2‑related factor 2 (Nrf2) signaling pathway and ferroptosis were assessed using western blotting analysis and an iron assay kit. TNFAIP1 expression was significantly upregulated in oxygen glucose deprivation and reperfusion (OGD/R)‑injured PC12 cells. However, knocking down TNFAIP1 expression restored PC12 cell viability and decreased apoptosis following OGD/R‑challenge. Furthermore, TNFAIP1 silencing significantly suppressed OGD/R‑induced oxidative stress and inflammatory damage in PC12 cells. TNFAIP1 knockdown inhibited ferroptosis via activation of the Nrf2 signaling pathway in OGD/R‑injured PC12 cells. Erastin treatment reversed the beneficial effects of TNFAIP1 knockdown on PC12 cell viability, apoptosis alleviation, oxidative stress and inflammation following OGD/R treatment. These results suggested that TNFAIP1 knockdown could alleviate OGD/R‑induced neuronal cell damage by suppressing Nrf2‑mediated ferroptosis, which might lay the foundation for the investigation of targeted‑therapy for cerebral I/R injury in clinic.

Introduction

Stroke was reported to cause 3.94 million (95% uncertainty interval 3.43-4.58) new cases in China in 2019(1). It has been previously reported that ischemic stroke constitutes >80% of all cases of stroke (2). This disease has become a key threat to public health with high rates of disability and mortality (3). At present, the primary therapeutic method for ischemic stroke is to recanalize occluded arteries through thrombolysis or thrombectomy (4). However, it has been reported that the restoration and reperfusion of blood flow into previously blood-deficient area has the potential to aggravate brain tissue injury and is frequently accompanied by cerebral ischemia-reperfusion (I/R) injury (CIRI) (5). CIRI is a dynamic and complex pathophysiological process, which is caused by a range of cellular and external physiological factors (6). Intracellular Ca2+ overload, overproduction of free radicals, excitatory amino acids, inflammatory cascade activation, acidosis, increased mitochondrial permeability and cell apoptosis are reported to be the primary factors of CIRI (7). Although thrombolysis and embolectomy restore blood flow to the infarcted brain tissue, such therapy also results in I/R (8,9). Therefore, it is important to elucidate the molecular mechanisms underlying CIR to develop novel strategies with significant efficacy for treatment of CIR.

TNFα-induced protein 1 (TNFAIP1), which is also called B12 or BACURD2, has numerous biological functions (10). It has been reported that TNFAIP1 is a key mediator of inflammation by activating NF-κB activity (11). Furthermore, TNFAIP1 has been reported to stimulate DNA polymerase δ activity and interact with proliferating cell nuclear antigen, which suggests that TNFAIP1 regulates the inflammatory response, cell proliferation and cell cycle progression (12). TNFAIP1 may also promote neurotoxicity (13). Gladwyn-Ng et al (14) previously reported that TNFAIP1 hinders neuronal migratory capabilities in the embryonic cortex and changes the morphology of the immature neuron (14). Another recent study reported that TNFAIP1 expression is upregulated following myocardial I/R and that TNFAIP1 knockdown ameliorates myocardial I/R injury (MIRI) via the Akt/GSK-3β/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway (15). However, the role of TNFAIP1 in CIR and its underlying mechanism remains poorly understood. Therefore, the present study evaluated the effects of TNFAIP1 on CIR and investigated how TNFAIP1 may regulate the pathophysiological process of CIRI.

Materials and methods

Cell culture and treatment

The rat adrenal gland cancer PC12 cell line was purchased from BioVector NTCC. DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (HyClone; Cytiva) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) were used to cultivate the cells at 37˚C with 5% CO2 for 2 h. To establish the oxygen glucose deprivation and reperfusion (OGD/R) model in vitro, PC12 cells were cultured in glucose-free DMEM (Procell Life Science & Technology Co., Ltd.) in an oxygen-free incubator supplied with 5% CO2 and 95% N2 at 37˚C for 2 h. Following hypoxia treatment, the media was replaced with normoxic glucose-containing medium and cells were transferred to an incubator supplied with 95% air and 5% CO2 at 37˚C for 24 h. Subsequently, cells were treated with 5 µM ML385 at room temperature for 6 h or 0.75 µM Erastin at room temperature for 24 h.

Cell transfection

For knockdown of TNFAIP1 expression, short hairpin RNAs (shRNAs) targeting TNFAIP1 with a pRNAU6.1 vector backbone (sh-TNFAIP1#1, 5'-GGAAGTGCTGACCGACAAA-3'; sh-TNFAIP1#2, 5'-GATTGCAGATAGCTAGCTA-3') and appropriate scrambled sequence negative control (sh-NC, 5'-GGTACGCAATAGGAGTGTGTG-3') were synthesized by Shanghai GenePharma Co., Ltd. The transfection of 100 nM of recombinants into PC12 cells was performed at 37˚C for 48 h using Lipofectamine® 2000 reagent (Thermo Fisher Scientific, Inc.). The transfection of cells with sh-TNFAIP1 was performed 24 h prior to OGD/R treatment. After 48 h, the cells were collected for subsequent experiments.

Cell Counting Kit-8 (CCK-8) assay

Following OGD/R treatment, cells were seeded into 96-well plates at a density of 5x103 cells and cultivated in DMEM with 10% FBS (HyClone; Cytiva) at room temperature for 24 h. Each well was treated with 10 µl CCK-8 solution (Sangon Biotech Co., Ltd.) added to each well for further incubation at 37˚C with 5% CO2 for 4 h. A microplate reader was used for the assessment of the optical density at 450 nm.

Flow cytometry

A total of 200 µl PC12 cells were rinsed with 1 ml pre-cold PBS twice and centrifuged. Cells were then resuspended into 100 µl binding buffer. Following incubation with 5 µl Annexin V-FITC on ice for 15 min, cells were stained with 10 µl propidium iodide (10 mg/ml) at 4˚C for 30 min in the dark. A flow cytometer (BD Biosciences) was used for detection. Flowjo vX.0.7 software (FlowJo LLC) was used to assess the rates of apoptosis.

Reactive oxygen species (ROS) detection

ROS Assay kit containing DCFH-DA (MilliporeSigma) was used to evaluate ROS generation. PC12 cells were probed using DCFH-DA (10 µM) at 37˚C in the dark for 30 min. Subsequently, PBS-rinsed cells were imaged using an Axio Observer D1fluorescence microscope (Carl Zeiss AG; magnification, x200).

Assessment of oxidative stress markers

PC12 cells were inoculated into six-well plates at a density of 4x105/well. Following aforementioned treatment, levels of intracellular glutathione (GSH) and malondialdehyde (MDA) were assessed using GSH Assay kit (cat. no. S0073; Beyotime Institute of Biotechnology) and MDA Assay Kit (cat. no. S0131S; Beyotime Institute of Biotechnology) according to the manufacturer's protocol. A microplate reader was used for colorimetric analysis.

ELISA

Following aforementioned treatment, the protein expression levels of inflammatory factors TNF-α, IL-1β and IL-6 in cell supernatant were assessed using corresponding ELISA kits (cat. nos. EK0526, EK0393 and EK0412, respectively; all Wuhan Boster Biological Technology, Ltd.). A microplate reader was used to assess the absorbance at 450 nm.

Assessment of Fe2+ levels

Following 48-h transfection, exposure of cells to OGD/R treatment with or without 5 µM ML385 was performed. Iron Assay kit (cat. no. ab83366; Abcam) was used to assess levels of Fe2+ in PC12 cells according to the manufacturer's protocol. A microplate reader was used to assess the absorbance at 593 nm of each well.

Reverse transcription-quantitative PCR (RT-qPCR)

RNA was isolated from cells using RNAiso Plus (Takara Bio, Inc.) and quantified using QuickDrop (Molecular Devices LLC) at 260 and 280 nm. cDNA was produced using PrimeScript™ RT Master Mix (Takara Bio, Inc.) before qPCR using SYBR® Premix Ex Taq™ II kit (Takara Bio, Inc.) using an ABI 7500 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The following thermocycling conditions were used for qPCR: 95˚C for 10 min; followed by 40 cycles of 95˚C for 10 sec and 60˚C for 60 sec. The primer sequences used for PCR were as follows: TNFAIP1 forward (F), 5'-ATCATCATCTTGCCTGGCCC-3' and reverse (R), 5'-GAACAAAGCTGTTCCCGTGC-3' and GAPDH F, 5'-GTCGTGGAGTCTACTGGCGTCTTCA-3' and R, 5'-TCGTGGTTCACACCCATCACAAACA-3'. GAPDH was used as an internal reference and the 2-ΔΔCq method (16) was used for the calculation of relative mRNA expression levels.

Western blotting

The proteins were quantified using a BCA protein assay kit (Thermo Fisher Scientific, Inc.) after the extraction of total proteins from the indicated PC12 cells using RIPA lysis buffer reagent (Beijing Solarbio Science & Technology Co., Ltd.). Following the separation of protein (30 µg/lane) using 10% SDS-PAGE, the proteins were transferred onto PVDF membranes. Membranes were blocked using 5% non-fat milk at room temperature for 2 h. Overnight incubation of membranes was performed at 4˚C with primary antibodies, supplied by Abcam, as follows: TNFAIP1 (1:1,000; cat. no. ab86934), Bcl-2 (1:1,000; cat. no. ab196495), Bax (1:1,000; cat. no. ab32503), cleaved caspase 3 (1:5,000; cat. no. ab214430), Nrf2 (1:1,000; cat. no. ab92946), Lamin B1 (1:1,000; cat. no. ab16048), heme oxygenase-1 (HO-1; 1:1,000; cat. no. ab68477), NADPH quinone dehydrogenase 1 (NQO-1; 1:1,000; cat. no. ab80588), GSH peroxidase 4 (GPX4; 1:1,000; cat. no. ab125066), ferritin heavy chain (1:1,000; cat. no. ab183781), ferroportin (FPN; 1:1,000; cat. no. ab239511), transferrin receptor 1 (TFR1; 1:1,000; cat. no. ab84036) and GAPDH (1:1,000; cat. no. ab8245). The membranes were washed with PBS for three times and then incubated with the HRP-labeled rabbit anti-mouse secondary antibodies (1:2,000; cat. no. ab6728; Abcam) at room temperature for 2 h. The antibody-labeled proteins were visualized using the ECL Detection Reagent (Shanghai Yeasen Biotechnology Co., Ltd.) and analyzed using ImageJ (version 1.49; National Institutes of Health).

Statistical analysis

All the experiments should be repeated at least three times. Data are presented as the mean ± standard deviation and were analyzed using SPSS 22.0 software (IBM Corp.). Unpaired Student's t test or one-way ANOVA followed by Bonferroni's post hoc test were used for comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Downregulation of TNFAIP1 decreases OGD/R-induced PC12 cell injury

To evaluate the role of TNFAIP1 in OGD/R-insulted PC12 cells, TNFAIP1 mRNA and protein expression levels were assessed in PC12 cells. The mRNA and protein expression levels of TNFAIP1 were significantly increased in OGD/R-treated PC12 cells compared with those in the control group (Fig. 1A and B). TNFAIP1 expression was knocked down in PC12 cells by transfection with sh-TNFAIP1#1 and #2. RT-qPCR and western blotting were performed to assess transfection efficiency. It was revealed that the mRNA and protein expressions of TNFAIP1 were significantly reduced compared with those in sh-NC after transfection with sh-TNFAIP1 (Fig. 1C and D). sh-TNFAIP1#1 exhibited superior transfection efficiency; therefore, sh-TNFAIP1#1 was used for subsequent experiments and was referred to as sh-TNFAIP1 thereafter. OGD/R treatment significantly suppressed PC12 cell viability compared with the control, which was significantly reversed by TNFAIP1 silencing compared with sh-NC group (Fig. 1E). Furthermore, the proportion of apoptotic cells was significantly elevated after OGD/R treatment compared with the control and significantly reversed by TNFAIP1 knockdown compared with the sh-NC group (Fig. 1F and G). Consistently, OGD/R significantly decreased Bcl-2 protein expression levels while significantly increasing those of Bax and cleaved caspase 3 compared with the control. However, TNFAIP1 knockdown significantly reversed the effects of OGD/R on expression levels of these proteins in PC12 cells (Fig. 1H).

TNFAIP1 silencing alleviates oxidative stress and the inflammatory response in OGD/R-induced PC12 cells

To assess the impact of TNFAIP1 knockdown in PC12 cells following OGD/R, the levels of oxidative stress and inflammation were evaluated. OGD/R significantly elevated levels of ROS compared with the control whereas TNFAIP1 silencing significantly decreased this enhancement compared with the sh-NC group (Fig. 2A and B). OGD/R treatment significantly decreased GSH but significantly increased MDA protein expression levels in PC12 cells compared with the control; however, knocking down TNFAIP1 expression significantly reversed OGD/R-induced oxidative stress compared with the sh-NC group (Fig. 2C and D). Furthermore, protein expression levels of TNF-α, IL-6 and IL-1β were significantly elevated following OGD/R compared with the control, which was significantly reversed after TNFAIP1 knockdown compared with the sh-NC group (Fig. 2E-G).

Knocking down TNFAIP1 expression suppresses ferroptosis via activation of the Nrf2 signaling pathway in OGD/R-injured PC12 cells

The potential mechanism by which TNFAIP1 regulated OGD/R-induced PC12 cells was assessed. Results obtained from western blotting demonstrated that OGD/R treatment significantly decreased cytoplasmic (C)-Nrf2 protein expression levels but significantly elevated those of nuclear (N)-Nrf2 and NQO-1 and markedly elevated those of HO-1 compared with the control. TNFAIP1 silencing significantly decreased C-Nrf2 protein expression levels further whereas it significantly increased those of N-Nrf2, HO-1 and NQO-1 compared with sh-NC group (Fig. 3A). To assess the association between the Nrf2 signaling pathway and ferroptosis downstream of TNFAIP1 in PC12 cells induced with OGD/R, the Nrf2 inhibitor ML385 was used to treat cells. OGD/R resulted in significantly elevated Fe2+ levels compared with the control and transfection with sh-TNFAIP1 significantly reversed the enhancement in Fe2+ levels that resulted from OGD/R compared with the sh-NC group. However, ML385 treatment significantly reversed the inhibitory effects of sh-TNFAIP1 on levels of Fe2+ in PC12 cells compared with the sh-TNFAIP1 group. It was also demonstrated that protein expression levels of GPX4, FTH1 and FPN were significantly decreased whilst those of TFR1 were significantly increased by OGD/R compared with the control and this was significantly reversed by TNFAIP1 silencing compared with the sh-NC group. The effects of TNFAIP1 knockdown on protein expression levels of GPX4, FTH1 and TFR1 were significantly reversed and the effect on the protein expression levels of FPN was markedly reversed by ML385 treatment compared with sh-TNFAIP1 group (Fig. 3C).

Erastin reverses the impact of TNFAIP1 silencing on OGD/R-induced PC12 cell injury

To assess the role of ferroptosis in TNFAIP-mediated OGD/R injury, erastin was added to the PC12 cells as a ferroptosis promoter. Treatment with erastin significantly decreased viability of OGD/R-induced PC12 cells with TNFAIP1 expression knocked down compared with sh-TNFAIP1 group (Fig. 4A). Erastin significantly increased cell apoptosis after treatment with OGD/R and transfection with sh-TNFAIP1 compared with sh-TNFAIP1 group (Fig. 4B and C). Significantly decreased Bcl-2 protein expression levels and significantly increased protein expression levels of Bax and cleaved caspase 3 were observed in OGD/R-stimulated PC12 cells with TNFAIP1 knockdown after treatment with erastin compared with the sh-TNFAIP1 group (Fig. 4D).

Erastin counteracts the impact of TNFAIP1 knockdown on oxidative stress and inflammation in PC12 cells following OGD/R injury

Erastin treatment significantly increased the production of ROS in PC12 cells transfected with sh-TNFAIP1 following OGD/R injury compared with sh-TNFAIP1 group (Fig. 5A and B). Furthermore, co-treatment with erastin and sh-TNFAIP1 significantly decreased protein expression levels of GSH and significantly increased MDA protein expression levels compared with those in the sh-TNFAIP1 group (Fig. 5C and D). Moreover, ELISA demonstrated that erastin significantly increased the protein expression levels of TNF-α, IL-6 and IL-1β in OGD/R-stimulated PC12 cells transfected with sh-TNFAIP1 compared with sh-TNFAIP1 group (Fig. 5E-G).

Discussion

Evidence has demonstrated that the production of certain cytokines, inflammatory cell infiltration and ROS production occur in ischemic injury, which can exacerbate the damage due to cerebral ischemia (17,18). When reperfusion occurs, recovery of blood supply may result in ROS production and inflammation, inducing CIRI (19). Therefore, novel effective therapeutic targets are required for treatment for ischemic stroke. In the present study, TNFAIP1 knockdown was demonstrated to increase cell viability and decrease oxidative stress and inflammation, in addition to suppressing ferroptosis in PC12 cells insulted with OGD/R, via the Nrf2 signaling pathway. This indicated the potential to use TNFAIP1 for the attenuation of CIRI.

OGD/R has been frequently used in research for induction of CIRI and has been reported to stimulate inflammation, apoptosis, autophagy and endoplasmic reticulum stress in cultured cortical neurons (20,21). In the present study, the OGD/R model was established to simulate CIRI in vitro. OGD/R stimulation was demonstrated to significantly suppress cell viability, aggravate apoptosis and oxidative stress and induce inflammatory damage in PC12 cells. TNFAIP1 is an evolutionarily conserved single-copy gene in humans, mice, rats and nematode worm that was first reported in umbilical vein endothelial cells (22). TNFAIP1 has been reported to serve a key role in neurodevelopment and a number of neurological diseases (23). For example, TNFAIP1 is overexpressed in the neurons of the cortex and hippocampus in the brains of APP/PS1 mice and the upregulation of TNFAIP1 can induce apoptosis in mice with Alzheimer's disease (24). A previous study reported that knocking down TNFAIP1 expression inhibits formaldehyde-induced neurotoxicity by suppression of cell apoptosis whilst increasing cell viability and neurite outgrowth by the inhibition of the AKT/cAMP response element binding protein (CREB) signaling pathway (25). Qiu et al (26) reported that TNFAIP1 knockdown elevates cell viability and suppresses apoptosis to prevent di(2-ethylhexyl) phthalate-induced neurotoxicity by triggering the CREB signaling pathway. In the present study, TNFAIP1 mRNA and protein expression levels were significantly upregulated in PC12 cells following OGD/R. TNFAIP1 silencing significantly reversed the OGD/R-induced decrease in cell viability while reducing cell apoptosis. Furthermore, oxidative stress and inflammatory response caused by OGD/R induction were attenuated by TNFAIP1 knockdown, which supported the protective effects of TNFAIP1 knockdown in PC12 cells following OGD/R induction. TNFAIP1 has been reported as being induced by TNF-α (27); however, in the present study it was demonstrated that the inhibition of TNFAIP1 significantly reduced TNF-α protein expression levels. This may be linked to previous reports that OGD/R can cause TNFAIP1 to regulate TNF-α in turn (15,28). Furthermore, Yi et al (25) reported that clearance of ROS suppresses formaldehyde-mediated upregulation of TNFAIP1 expression; however in the present study, it was demonstrated that TNFAIP1-silencing inhibited production of ROS in OGD/R-treated PC12 cells.

It has been previously reported that the Nrf2 signaling pathway is associated with I/R process in numerous types of tissue (29,30). Zhao et al (31) reported that sulforaphane attenuates liver injury from intestinal I/R via the Nrf2/antioxidant response element pathway. Wei et al (32) reported that Nrf2 protects against neuronal and capillary degeneration following retinal I/R injury. Furthermore, another study reported that TNFAIP1 expression is increased following I/R injury and TNFAIP1 knockdown alleviates MIRI via the AKT/GSK3β/Nrf2 signaling pathway (15). Therefore, it was hypothesized that TNFAIP1 is involved in neuron injury from CIRI. In the present study, C-Nrf2 protein expression levels were significantly decreased and N-Nrf2 protein expression levels were significantly enhanced following OGD/R treatment, whereas transfection with sh-TNFAIP significantly decreased C-Nrf2 protein expression levels and significantly increased protein expression levels of N-Nrf2 further. It can be hypothesized that excessive oxidative stress due to OGD/R promoted separation of Kelch-like ECH-associated protein 1 from Nrf-2 to activate Nrf2 and that silencing of TNFAIP1 expression may prolong activation of the Nrf2 signaling pathway following OGD/R. The Nrf2 signaling pathway is a key pathway that leads to ferroptosis (33). Yuan et al (34) reported that kaempferol inhibits OGD/R-induced ferroptosis in neurons via the AKT/Nrf2/GPX4 signaling pathway. The present study demonstrated that OGD/R significantly enhanced levels of Fe2+ and TFR1 protein expression levels but significantly diminished protein expression levels of GPX4, FTH1 and FPN. TNFAIP1 silencing significantly reversed this trend and ML385 treatment prevented this reversal. Erastin was added to the PC12 cells to promote ferroptosis, which demonstrated that erastin reversed the beneficial effects of TNFAIP1 on viability, oxidative stress and inflammatory damage in OGD/R-induced PC12 cells. These results suggested that downregulation of TNFAIP1 expression may alleviate OGD/R-induced neuronal cell damage by inhibition of ferroptosis via regulation of the Nrf2 signaling pathway.

In summary, the present study demonstrated the potential protective role of TNFAIP1 silencing in OGD/R-injured neurocytes and the key role of Nrf2-mediated ferroptosis in the viability, oxidative stress, apoptosis and inflammatory damage of PC12 cells following OGD/R stimulation, which suggested that TNFAIP1 may be a promising therapeutic target for CIRI. However, there are also some limitations of the present study. First, the function of TNFAIP1 in CIRI in a clinical setting was not explored. Second, an in vivo CIRI model was not established to investigate the role of TNFAIP1 in CIRI.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Basic Public Welfare Research Program of Zhejiang Province, China (grant no. LGF20H270003).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LX and KL designed the present study and drafted and revised the manuscript. LX, JZ, HS, GZ and XJ performed the experiments. ML and PZ reviewed the literature and analyzed the data. LX and KL confirmed the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ma Q, Li R, Wang L, Yin P, Wang Y, Yan C, Ren Y, Qian Z, Vaughn MG, McMillin SE, et al: Temporal trend and attributable risk factors of stroke burden in China, 1990-2019: An analysis for the global burden of disease study 2019. Lancet Public Health. 6:e897–e906. 2021.PubMed/NCBI View Article : Google Scholar

2 

Grysiewicz RA, Thomas K and Pandey DK: Epidemiology of ischemic and hemorrhagic stroke: incidence, prevalence, mortality, and risk factors. Neurol Clin. 26:871–895. 2008.PubMed/NCBI View Article : Google Scholar

3 

Owens B: Stroke. Nature. 510(S1)2014.PubMed/NCBI View Article : Google Scholar

4 

Pan J, Konstas AA, Bateman B, Ortolano GA and Pile-Spellman J: Reperfusion injury following cerebral ischemia: Pathophysiology, MR imaging, and potential therapies. Neuroradiology. 49:93–102. 2007.PubMed/NCBI View Article : Google Scholar

5 

Huang L, Chen C, Zhang X, Li X, Chen Z, Yang C, Liang X, Zhu G and Xu Z: Neuroprotective effect of curcumin against cerebral ischemia-reperfusion via mediating autophagy and inflammation. J Mol Neurosci. 64:129–139. 2018.PubMed/NCBI View Article : Google Scholar

6 

Wang H, Chen S, Zhang Y, Xu H and Sun H: Electroacupuncture ameliorates neuronal injury by Pink1/Parkin-mediated mitophagy clearance in cerebral ischemia-reperfusion. Nitric Oxide. 91:23–34. 2019.PubMed/NCBI View Article : Google Scholar

7 

Yang J, Chen M, Cao RY, Li Q and Zhu F: The role of circular RNAs in cerebral ischemic diseases: Ischemic stroke and cerebral ischemia/reperfusion injury. Adv Exp Med Biol. 1087:309–325. 2018.PubMed/NCBI View Article : Google Scholar

8 

Moussaddy A, Demchuk AM and Hill MD: Thrombolytic therapies for ischemic stroke: Triumphs and future challenges. Neuropharmacology. 134:272–279. 2018.PubMed/NCBI View Article : Google Scholar

9 

Gobin YP, Starkman S, Duckwiler GR, Grobelny T, Kidwell CS, Jahan R, Pile-Spellman J, Segal A, Vinuela F and Saver JL: MERCI 1: A phase 1 study of mechanical embolus removal in cerebral ischemia. Stroke. 35:2848–2854. 2004.PubMed/NCBI View Article : Google Scholar

10 

Zhang CL, Wang C, Yan WJ, Gao R, Li YH and Zhou XH: Knockdown of TNFAIP1 inhibits growth and induces apoptosis in osteosarcoma cells through inhibition of the nuclear factor-κB pathway. Oncol Rep. 32:1149–1155. 2014.PubMed/NCBI View Article : Google Scholar

11 

Zhao Y, Li S, Xia N, Shi Y and Zhao CM: Effects of XIST/miR-137 axis on neuropathic pain by targeting TNFAIP1 in a rat model. J Cell Physiol. 233:4307–4316. 2018.PubMed/NCBI View Article : Google Scholar

12 

Yang L, Liu N, Hu X, Zhang W, Wang T, Li H, Zhang B, Xiang S, Zhou J and Zhang J: CK2 phosphorylates TNFAIP1 to affect its subcellular localization and interaction with PCNA. Mol Biol Rep. 37:2967–2973. 2010.PubMed/NCBI View Article : Google Scholar

13 

Liu N, Yu Z, Xun Y, Li M, Peng X, Xiao Y, Hu X, Sun Y, Yang M, Gan S, et al: TNFAIP1 contributes to the neurotoxicity induced by Aβ25-35 in Neuro2a cells. BMC Neurosci. 17(51)2016.PubMed/NCBI View Article : Google Scholar

14 

Gladwyn-Ng IE, Li SS, Qu Z, Davis JM, Ngo L, Haas M, Singer J and Heng JI: Bacurd2 is a novel interacting partner to Rnd2 which controls radial migration within the developing mammalian cerebral cortex. Neural Dev. 10(9)2015.PubMed/NCBI View Article : Google Scholar

15 

Wen L, Yang QH, Ma XL, Li T, Xiao S and Sun CF: Inhibition of TNFAIP1 ameliorates the oxidative stress and inflammatory injury in myocardial ischemia/reperfusion injury through modulation of Akt/GSK-3β/Nrf2 pathway. Int Immunopharmacol. 99(107993)2021.PubMed/NCBI View Article : Google Scholar

16 

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

17 

Zhao J, Wang H, Dong L, Sun S and Li L: miRNA-20b inhibits cerebral ischemia-induced inflammation through targeting NLRP3. Int J Mol Med. 43:1167–1178. 2019.PubMed/NCBI View Article : Google Scholar

18 

Pei H, Song X, Peng C, Tan Y, Li Y, Li X, Ma S, Wang Q, Huang R, Yang D, et al: TNF-α inhibitor protects against myocardial ischemia/reperfusion injury via Notch1-mediated suppression of oxidative/nitrative stress. Free Radic Biol Med. 82:114–121. 2015.PubMed/NCBI View Article : Google Scholar

19 

Hao MQ, Xie LJ, Leng W and Xue RW: Trim47 is a critical regulator of cerebral ischemia-reperfusion injury through regulating apoptosis and inflammation. Biochem Biophys Res Commun. 515:651–657. 2019.PubMed/NCBI View Article : Google Scholar

20 

Qin H, Tan W, Zhang Z, Bao L, Shen H, Wang F, Xu F and Wang Z: 15d-prostaglandin J2 protects cortical neurons against oxygen-glucose deprivation/reoxygenation injury: Involvement of inhibiting autophagy through upregulation of Bcl-2. Cell Mol Neurobiol. 35:303–312. 2015.PubMed/NCBI View Article : Google Scholar

21 

Liu Y, Qu X, Yan M, Li D and Zou R: Tricin attenuates cerebral ischemia/reperfusion injury through inhibiting nerve cell autophagy, apoptosis and inflammation by regulating the PI3K/Akt pathway. Hum Exp Toxicol. 41(9603271221125928)2022.PubMed/NCBI View Article : Google Scholar

22 

Wolf FW, Marks RM, Sarma V, Byers MG, Katz RW, Shows TB and Dixit VM: Characterization of a novel tumor necrosis factor-alpha-induced endothelial primary response gene. J Biol Chem. 267:1317–1326. 1992.PubMed/NCBI

23 

Liu Y, Sun H and Sun Y: LncRNA p21, downregulating miR-181b, aggravates neuropathic pain by upregulating Tnfaip1 and inhibit the AKT/CREB axis. Brain Res Bull. 171:150–161. 2021.PubMed/NCBI View Article : Google Scholar

24 

Xiao Y, Li Y, Zhang H, Yang L, Jiang Y, Wei C, Feng X, Xun Y, Yuan S, Xiang S and Liu N: TNFAIP1 is upregulated in APP/PS1 mice and promotes apoptosis in SH-SY5Y cells by binding to RhoB. J Mol Neurosci. 71:1221–1233. 2021.PubMed/NCBI View Article : Google Scholar

25 

Yi J, Zhu M, Qiu F, Zhou Y, Shu P, Liu N, Wei C and Xiang S: TNFAIP1 mediates formaldehyde-induced neurotoxicity by inhibiting the Akt/CREB pathway in N2a cells. Neurotox Res. 38:184–198. 2020.PubMed/NCBI View Article : Google Scholar

26 

Qiu F, Zhou Y, Deng Y, Yi J, Gong M, Liu N, Wei C and Xiang S: Knockdown of TNFAIP1 prevents di-(2-ethylhexyl) phthalate-induced neurotoxicity by activating CREB pathway. Chemosphere. 241(125114)2020.PubMed/NCBI View Article : Google Scholar

27 

Tang X, Tangkham T, Aljahdali B, Lee S, Su M and Dibart S: The role of TNF-α induced protein 1 in the activation of pro-apoptotic proteins. Hum Cell. 34:1123–1129. 2021.PubMed/NCBI View Article : Google Scholar

28 

Aljahdali BH: Regulation of TNF-α gene expression by TNFAIP1 as an activator or suppressor in response to lipopolysaccharide (unpublished PhD thesis). Boston University, 2019.

29 

Xiao C, Xia ML, Wang J, Zhou XR, Lou YY, Tang LH, Zhang FJ, Yang JT and Qian LB: Luteolin attenuates cardiac ischemia/reperfusion injury in diabetic rats by modulating Nrf2 antioxidative function. Oxid Med Cell Longev. 2019(2719252)2019.PubMed/NCBI View Article : Google Scholar

30 

He M, Pan H, Chang RC, So KF, Brecha NC and Pu M: Activation of the Nrf2/HO-1 antioxidant pathway contributes to the protective effects of Lycium barbarum polysaccharides in the rodent retina after ischemia-reperfusion-induced damage. PLoS One. 9(e84800)2014.PubMed/NCBI View Article : Google Scholar

31 

Zhao HD, Zhang F, Shen G, Li YB, Li YH, Jing HR, Ma LF, Yao JH and Tian XF: Sulforaphane protects liver injury induced by intestinal ischemia reperfusion through Nrf2-ARE pathway. World J Gastroenterol. 16:3002–3010. 2010.PubMed/NCBI View Article : Google Scholar

32 

Wei Y, Gong J, Yoshida T, Eberhart CG, Xu Z, Kombairaju P, Sporn MB, Handa JT and Duh EJ: Nrf2 has a protective role against neuronal and capillary degeneration in retinal ischemia-reperfusion injury. Free Radic Biol Med. 51:216–224. 2011.PubMed/NCBI View Article : Google Scholar

33 

Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23(101107)2019.PubMed/NCBI View Article : Google Scholar

34 

Yuan Y, Zhai Y, Chen J, Xu X and Wang H: Kaempferol ameliorates oxygen-glucose deprivation/reoxygenation-induced neuronal ferroptosis by activating Nrf2/SLC7A11/GPX4 axis. Biomolecules. 11(923)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2023
Volume 25 Issue 1

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
Xiong L, Zhang J, Shi H, Zhu G, Ji X, Li M, Zhu P and Luo K: Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis. Exp Ther Med 25: 25, 2023
APA
Xiong, L., Zhang, J., Shi, H., Zhu, G., Ji, X., Li, M. ... Luo, K. (2023). Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis. Experimental and Therapeutic Medicine, 25, 25. https://doi.org/10.3892/etm.2022.11724
MLA
Xiong, L., Zhang, J., Shi, H., Zhu, G., Ji, X., Li, M., Zhu, P., Luo, K."Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis". Experimental and Therapeutic Medicine 25.1 (2023): 25.
Chicago
Xiong, L., Zhang, J., Shi, H., Zhu, G., Ji, X., Li, M., Zhu, P., Luo, K."Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis". Experimental and Therapeutic Medicine 25, no. 1 (2023): 25. https://doi.org/10.3892/etm.2022.11724